You are on page 1of 11

Cell Host & Microbe

Review

The Impact of Dietary Fiber on Gut


Microbiota in Host Health and Disease
€ckhed1,4,*
Kassem Makki,1 Edward C. Deehan,2 Jens Walter,2,3 and Fredrik Ba
1Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska

University Hospital, Gothenburg, Sweden


2Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
3Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada
4Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of

Health Sciences, University of Copenhagen, Copenhagen, Denmark


*Correspondence: fredrik@wlab.gu.se
https://doi.org/10.1016/j.chom.2018.05.012

Food is a primordial need for our survival and well-being. However, diet is not only essential to maintain
human growth, reproduction, and health, but it also modulates and supports the symbiotic microbial com-
munities that colonize the digestive tract—the gut microbiota. Type, quality, and origin of our food shape
our gut microbes and affect their composition and function, impacting host-microbe interactions. In this
review, we will focus on dietary fibers, which interact directly with gut microbes and lead to the production
of key metabolites such as short-chain fatty acids, and discuss how dietary fiber impacts gut microbial ecol-
ogy, host physiology, and health. Hippocrates’ notion ‘‘Let food be thy medicine and medicine be thy food’’
remains highly relevant millennia later, but requires consideration of how diet can be used for modulation of
gut microbial ecology to promote health.

Introduction colorectal cancer (CRC), allergies, autoimmune diseases, and


Our gut harbors trillions of microbes representing all kingdoms of obesity and its associated pathologies. These diseases can, at
life that are essential for host development and physiology. This least in part, be prevented by dietary fiber, arguing for attempts
‘‘gut microbiota’’ constitutes a complex community that inter- to close the ‘‘fiber gap’’ through adjustment of human diet (Dee-
acts with each other and with the host to modulate biological han and Walter, 2016). Here we will discuss how dietary fiber
processes essential for health. Our understanding of the biolog- impacts gut microbial ecology, host physiology, and health by
ical roles of the gut microbiome, which include modulating juve- specifically focusing on mechanisms by which a low-fiber diet
nile growth (Schwarzer et al., 2016), maturation of the immune disrupts the microbial ecosystem and leads to a predisposition
system (Rescigno, 2014), and modulation of glucose and lipid to chronic inflammatory diseases.
metabolism (Ba €ckhed et al., 2004), has increased dramatically
in the past decade. Dietary Fibers: Definitions, Characteristics, and Origin
The microbiome contributes to homeostatic regulation in The definition of dietary fiber has been a matter of debate and
different tissues in our body (Schroeder and Ba €ckhed, 2016). has evolved over the last decade (Jones, 2014). Most countries
However, although the overall interrelationship of humans with adopted the definition of the Codex Alimentarius commission
their microbiota can be considered a mutualistic symbiosis (Wal- from 2009, which defines dietary fiber as edible carbohydrate
ter et al., 2011), eubiosis, which refers to a healthy balance of the polymers with three or more monomeric units that are resistant
microbes in the gut, can be disrupted, leading to the develop- to the endogenous digestive enzymes and thus neither hydro-
ment of various chronic diseases with an underlying inflamma- lyzed nor absorbed in the small intestine, and that belong to
tory condition (Hand et al., 2016). Most microbiome-linked the following categories: (1) edible carbohydrate polymers natu-
pathologies have dramatically increased over the past century, rally occurring in foods such as fruits, vegetables, legumes, and
suggesting that a change in lifestyle might disrupt gut microbiota cereals; (2) edible carbohydrate polymers obtained from food
symbiosis due to the loss of beneficial, protective microbes raw materials by physical, enzymatic, and chemical means that
(Logan et al., 2016). In fact, Western-style diet, low in micro- have a proven physiological benefit; and (3) synthetic carbohy-
biota-accessible carbohydrates (MACs), may irreversibly reduce drate polymers with a proven physiological benefit. Although
microbial diversity and lead to the disappearance of specific most national authorities adhere to this definition, some differ-
bacterial species in the digestive system (Sonnenburg et al., ences are found between dietary fiber definitions, and they
2016). Accordingly, the low intake of dietary fibers and the mainly concern (1) considering some non-carbohydrates such
increased amounts of fat and sugar in our food, typical for a as lignin and other substances present in cell walls linked to
Westernized lifestyle and nutrition, may at least in part contribute polysaccharides as dietary fibers (Stephen et al., 2017) and (2)
to depletion of specific bacterial taxa (Sonnenburg and Sonnen- the minimum number of carbohydrate monomers to be included.
burg, 2014). These alterations may result in dysfunctions, Dietary fibers are classified according to several parameters,
contributing to the increase in the development of chronic including their primary food source, their chemical structure,
inflammatory diseases such as intestinal bowel disease (IBD), their water solubility and viscosity, and their fermentability.

Cell Host & Microbe 23, June 13, 2018 ª 2018 Elsevier Inc. 705
Cell Host & Microbe

Review

Dietary fibers are subdivided either into polysaccharides (non- observed between industrialized and non-industrialized human
starch polysaccharides [NSPs], resistant starch [RS], and resis- populations, suggesting that these bacterial populations are
tant oligosaccharides [ROs]) or into insoluble and soluble forms driven by long-term differences in diet, e.g., meat (drives Bacter-
(Deehan et al., 2017). Most insoluble forms such as cellulose oides in the West) and dietary fiber (drives Prevotella in non-West-
and hemi-cellulose have a fecal bulking effect, as they reach ernized populations). Comparing gut microbiota of children from
the colon and are not, or only slowly, digested by the gut bacte- rural (Burkina Faso [BF]) versus urban (Italy) communities
ria. Most soluble fibers do not contribute to fecal bulking, but are reflected the impact of diet on the microbiome. De Filippo et al.
fermented by the gut bacteria and thus give rise to metabolites found a significant difference within the gut microbiota composi-
such as short-chain fatty acids (SCFAs). In contrast to ROs, tion in both groups after weaning. A significant increase in the
most soluble NSPs, especially polymers with high molecular abundance of bacteria from the genera Prevotella and Xylani-
weight such as guar gum, certain pectins, b-glucans, and bacter associated with higher levels of fecal SCFAs was
psyllium, are viscous, meaning that they are able to form a gel observed in BF children, thus reflecting the ability of these individ-
structure in the intestinal tract that can delay absorption of uals to degrade complex carbohydrates (De Filippo et al., 2010).
glucose and lipids influencing post-prandial metabolism (Dee- These differences are independent of ethnicity as the gut bacte-
han et al., 2017). rial communities of BF children living in urban, as opposed to
Soluble and insoluble fibers are found in different food sources rural, areas of BF became more similar to the Italian children.
such as legumes, vegetables, nuts, seeds, fruits, and cereals in When people move to urban areas, they are exposed to a West-
different proportions. However, not all types of fibers are present ernized lifestyle, including access to food rich in fat and simple
in the same food categories. RS can only be found in starchy sugars. Accordingly, the microbiota of children living in the urban-
foods such as cereals, legumes, tubers, and non-mature fruits ized areas of BF have bacteria that are more suited to metabolize
like green bananas, while pectins are more enriched in fruits animal protein, fat, and sugar-rich foods, while children living in
and some vegetables, whereas b-glucans and arabinoxylans rural zones have a bacterial reservoir (enrichment in Prevotella,
are present in cereals (Lovegrove et al., 2017). Although fibers Treponema, and Succinivibrio) adapted for fiber and carbohy-
are present in a wide range of plant-based food sources, con- drate fermentation from vegetables. Interestingly, the microbial
sumption is low in Western countries. Fortification of foods communities of BF children living in urban areas were compara-
with extracted or synthesized non-digestible carbohydrates or ble to those of Italian children, highlighting the important impact
their use in dietary supplements therefore constitutes a strategy of the diet independent of host genetic (De Filippo et al., 2017).
to increase fiber intake. A wide range of these carbohydrate These findings are supported by a study from Schnorr et al.
polymers and oligosaccharides is commercially available (Dee- that showed that the Hadza hunter-gatherer individuals possess
han and Walter, 2016). Some of these compounds are consid- higher levels of microbial richness and biodiversity compared to
ered ‘‘prebiotics’’ on the premise that they exert health benefits Italian urban controls. The gut microbiota of these individuals
by selectively inducing beneficial bacterial populations in the gut. had increased Bacteroidetes and decreased Firmicutes abun-
However, the assignment of only certain fibers as prebiotics dances, and unexpectedly they almost lack the Actinobacteria
based on these criteria is somehow arbitrary (Bindels et al., phylum, including low levels of Bifidobacterium. Interestingly,
2015; Deehan et al., 2017). As we describe in the following sec- SCFA production in Hadza was characterized by increased
tions, virtually all fibers will induce specific shifts in microbiota propionate concentrations, while the Italian cohort had more
composition due to competitive interactions, and which of these butyrate (Schnorr et al., 2014). This segregation in SCFA produc-
compositional shifts contribute to health benefits, or if they are tion may reflect differences in dietary variation related to the
even functionally relevant, has not yet been established (Bindels amount and type of fiber and carbohydrates consumed by
et al., 2015). In contrast, the mechanisms that have been demon- both communities. Similarly, analysis of the microbiome of
strated to contribute to health benefits do not rely on a selective Malawian, Venezuelan, and American populations from infants
utilization of the carbohydrate but rather on metabolic com- to adults showed significant differences, which were associated
pounds (e.g., SCFAs) (Koh et al., 2016), physiological changes with diet and culture (Yatsunenko et al., 2012). The microbiome
(pH lowering), or protection of the mucus layer (Schroeder of Malawian and Venezuelan infants was characterized by the
et al., 2018; Zou et al., 2018). Therefore, it may be worth consid- enrichment of bacterial genes implicated in vitamin B12 biosyn-
ering a shift in focus of the prebiotic concept away from the thesis, host glycan utilization, and urea catabolism, while
selective effect of specific dietary components on gut microbial increased ability to metabolize fucose was observed in US
communities and instead toward ecological and functional con- infants. In the same study, significant differences in microbial
sequences of fiber fermentation, which is more relevant for host functions between US versus Malawian and Venezuelan adults
physiology (Bindels et al., 2015). were shown that corresponded to carnivorous (protein
consumption) versus herbivorous (dietary fiber consumption)
Impact of Dietary Fiber on Microbial Ecology in the Gut profiles, respectively.
Diet has a major impact on gut microbiota composition, diversity, Consistent with these observations, environmental lifestyle
and richness. Different components of the diet will shape the gut changes rather than host genetics influence gut microbiota
bacterial communities in a time-dependent manner. Long-term diversity, and industrialization leads to a significant depletion of
dietary patterns, particularly the intake of protein and animal fat species (Smits et al., 2017). The underlying mechanisms for
(Bacteroides) versus carbohydrates or plant-based foods (Prevo- reduced diversity are proposed to include antibiotic utilization,
tella), are associated with so-called enterotypes (Wu et al., 2011). clinical practices (e.g., cesarean sections), and sanitation (Son-
This dichotomy in the Prevotella/Bacteroides ratio was also nenburg and Sonnenburg, 2014). In agreement with this notion,

706 Cell Host & Microbe 23, June 13, 2018


Cell Host & Microbe

Review

the most diverse microbiome to date is observed in the Yano- sources of fibers depending on availability (Flint et al., 2012).
mami, an indigenous population with limited contact to the Species belonging to Firmicutes and Actinobacteria are the
industrialized world (Clemente et al., 2015; Smits et al., 2017). main responders to dietary fiber (Deehan et al., 2017), although
However, even in non-industrialized human populations with they contain relatively few fiber-metabolizing enzymes per
access to antibiotics, such as rural Papua New Guineans, diver- organism. However, they generally have more specialized roles
sity is higher than in the West (Martı́nez et al., 2015). Thus, other such as the initiation of complex substrate degradation. For
environmental factors that differentiate Western and non-West- example, administration of RS has been shown to enrich Bifido-
ern human populations such as diet may be more important. bacterium adolescentis, Ruminoccocus bromii, Eubacterium
The non-Western populations consume far less refined diets rectale, and Parabacteroides distasonis in a subset of individuals
and significantly higher proportions of dietary fibers, suggesting dependent on RS (Martı́nez et al., 2010; Walker et al., 2011). In
that this dietary component contributes to microbiota richness. contrast, the consumption of galactooligosaccharides mainly
In fact, Sonnenburg et al. investigated the consequences of induces Bifidobacterium species that possess the enzymatic
the lack of fiber (referred to as MACs) intake in mice colonized machinery to efficiently utilize this substrate (Davis et al.,
with a human microbiota and showed that a low-MAC diet led 2011). It is not only the enzymatic capacity (as a primary fiber
to dramatically reduced microbial diversity in just three genera- degrader) that determines the ability of a microbe to benefit
tions, which could not be restored when mice were moved to a from a dietary fiber, but also its ability to ‘‘adhere’’ to a substrate,
normal-MAC diet (Sonnenburg et al., 2016). Interestingly, they tolerate the environmental conditions changed through the fiber
also observed seasonal reductions in microbiome diversity in (e.g., increased acidity through fermentation), and benefit from
Hadza hunter-gatherer individuals of Tanzania. The microbiota carbohydrate breakdown products (secondary fiber degraders)
composition and function of this community varied according and metabolites (through cross-feeding) (Deehan et al., 2017).
to seasonal changes and reflected dietary habit as well as the Primary fiber degraders can hereby function as ‘‘keystone’’ spe-
type of food eaten (Smits et al., 2017). During the wet season, cies that initiate the utilization of a complex fiber through what
the representation of CAZymes in the metagenome that are spe- can be considered a ‘‘guild’’ of species (Zhao et al., 2018). For
cific for plant carbohydrates was reduced and was associated example, R. bromii is considered a keystone species for the
with a disappearance-reappearance cycle of specific OTUs degradation of RS and contributes significantly to butyrate
(operational taxonomic units) such as Prevotellaceae, which production in the colon, although the species itself does not pro-
are lost in industrialized populations. Similar seasonal observa- duce butyrate. Similar keystone species are likely to exist for
tions were found in hibernating free-ranging brown bears in other dietary fiber types but have not yet been identified.
which microbes associated with fiber utilization were increased The impact of dietary fiber on microbiota composition displays
during the late spring and depleted during hibernation. The several consistent characteristics. First, the observed shifts
enrichment of these bacteria led to increased efficiency in calorie induced by non-digestible carbohydrates in humans, regardless
harvest from a fiber-rich diet (Sommer et al., 2016). Thus, peri- if they are accepted prebiotics or not, are restricted to a limited
odic reduction in fiber intake does not appear to have long-last- number of taxa (Davis et al., 2011; Martı́nez et al., 2010; Walker
ing effects on the microbiome (Smits et al., 2017), whereas long- et al., 2011). Second, the magnitude of the induced changes can
term reduction in fiber intake, like in the Western world, may lead be substantial, with specific species constituting more than 30%
to permanent extinction of important microbial taxa, similar to of the total sequences obtained by amplicon sequencing of the
the findings in mice (Sonnenburg et al., 2016). fecal microbiota (Davis et al., 2011; Martı́nez et al., 2010; Walker
In addition to the long-term effects of the diet on the micro- et al., 2011). However, these changes are only maintained so
biome as outlined above, gut microbial communities respond long as the substrate is consumed. Third, the microbial response
within 24 hr to dramatic changes in macronutrient composition to dietary fiber is highly individualized (Davis et al., 2011; Martı́-
(David et al., 2014). In particular, taxa with fiber-degrading nez et al., 2010). The reason for this individuality is not yet under-
capacity increased when humans were fed a plant-based diet. stood. Individuals may lack keystone species (Ze et al., 2012) or
In agreement, a dramatic and rapid rearrangement in the human lack strains that possess the enzymatic capacity to utilize a
microbiome within 24 hr after reduction of carbohydrate specific substrate (Zhao et al., 2018).
(including fiber) intake to 30 g/day was observed in a recent
human cohort study (Mardinoglu et al., 2018). As expected, the Microbial Metabolism of Dietary Fibers and Functional
loss in carbohydrates dramatically reduced the abundance of Implications
fiber-degrading bacteria, while the abundance of Lactococcus, Dietary fibers are important energy sources for cecum and
Eggerrthella, and Streptococcus increased, resulting in reduced colon-residing microbiota. Anaerobic bacteria under specific
levels of SCFAs (Mardinoglu et al., 2018). intestinal conditions activate their machinery, constituted of
key enzymes and metabolic pathways, which can metabolize
Fiber Forms the Food Webs for Bacteria complex carbohydrates, thus leading to the production of me-
Dietary administration of fiber alters the niche environment in the tabolites such as SCFAs.
gut by providing substrates for microbial growth, allowing micro- SCFAs are organic products mainly composed of acetate,
bial species that are able to utilize these substrates to expand propionate, and butyrate. SCFAs possess key roles in regulating
their populations (Deehan et al., 2017). Together, the gut micro- host metabolism, immune system, and cell proliferation (Koh
biome harbors 130 glycoside hydrolase, 22 polysaccharide et al., 2016). SCFAs are found at high concentration in the cecum
lyase, and 16 carbohydrate esterase families, which provide and proximal colon, where they are used as energy sources in
the microbiome flexibility to switch between different energy colonocytes (especially butyrate), but can also be transported

Cell Host & Microbe 23, June 13, 2018 707


Cell Host & Microbe

Review
Figure 1. Effect of Low- and High-Fiber Diet
on Gut Microbiota Composition, Diversity,
Prudent Diet and Function in Host Physiology
Western Diet A diet rich in fiber contributes to the maintenance of
a healthy gut microbiota associated with increased
diversity and functions such as the production of
short-chain fatty acids (SCFAs). With the industri-
alization of the diet, low fiber intake, and high protein
and sugar consumption, the diversity of the gut
bacteria is reduced and their function is altered,
including significant reduction in their ability to
produce SCFAs, and associated with the appear-
High fat ance of chronic inflammatory diseases. High fiber
Phytochemicals Omega-3 fatty acids High sugar
intake and the production of SCFAs by the gut
Emulsifiers
bacteria enhance mucus and anti-microbial peptide
High dietary fiber, High meat protein production, and increase expression of tight junc-
MACs, Prebiotics Low dietary fiber tion proteins. In addition, SCFAs reduce oxygen
levels and maintain a functional immune system.
Toxic metabolites, Toxic
These biological processes are disrupted when the
SCFAs BCFAs bile acids diet is shifted toward a Western lifestyle and may
lead to increased susceptibility to infections and
Hyperglycemia
IBD, and to impaired physiology.

Enhanced mucus Increased antimicrobial Enhanced mucus Reduced antimicrobial


secretion peptides degradation peptides
CRC (Windey et al., 2012). Given the trade-
Lumen off between saccharolytic and proteolytic
fermentation, a high-fiber diet is likely to
inhibit protein fermentation counteracting
many of the detrimental effects of meat
Host
and fat, making these food components
Regulated immune response Proinflammatory immune response less detrimental.
upregulated downregulated
oxygen oxygen
tight junctions tight junctions
Effect of Dietary Fibers and SCFAs
Functional intestinal barrier on Host
Dysfunctional intestinal barrier

Immune Functional Dysfunctional Chronic


Mucus Production and Luminal
Health Inflammation
homeostasis barrier barrier disease Oxygen Levels
The gut epithelium is covered and pro-
Dietary fiber degrading Mucus degrading tected by a mucus layer, thus keeping
gut microbiota gut microbiota
bacteria separated from the mucosa
(Johansson et al., 2008). One of the
mechanisms used by the host to prevent
microbe invasions and susceptibility to
to the peripheral circulation via the portal vein to act on the liver infections is to maintain a well-structured and intact mucus
and peripheral tissues. Although the levels of SCFAs are low in layer. The gut microbiota and diet are two important compo-
the peripheral circulation, it is now well accepted that they act nents to maintain a normal structure and production of the
as signaling molecules and regulate different biological pro- intestinal mucus. An altered gut microbiota resulting from a
cesses in the host (Koh et al., 2016). diet low in fibers leads to a severe deterioration of the mucus
A low intake of dietary fiber does not only lead to reduced layer and can enhance the susceptibility to infections and the
microbial diversity and SCFA production, but also shifts the development of chronic inflammatory diseases (Figure 1) (De-
gut microbial metabolism toward the utilization of less favorable sai et al., 2016; Johansson et al., 2008; Schroeder et al.,
substrates, particularly dietary and endogenously supplied pro- 2018; Zou et al., 2018).
teins (Cummings and Macfarlane, 1991) and host mucins (Desai Dietary fibers and SCFAs stimulate mucus production and
et al., 2016; Schroeder et al., 2018; Zou et al., 2018), which may secretion. Both acetate and butyrate maintain a balance for
be detrimental to the host. Supplying human volunteers with a mucus production and secretion. Bacteroides thetaiotaomicron,
high-protein, low-carbohydrate diet did not only significantly an acetate and propionate producer, promotes goblet cell differ-
reduce the production of total SCFAs and butyrate (Duncan entiation and expression of mucin-related genes. In contrast,
et al., 2007), but also led to an increase in potentially detrimental Faecalibacterium prausnitzii, a consumer of acetate and a buty-
metabolites derived from the fermentation of amino acids, rate producer, reduces the effect of acetate on mucus and
including branched-chain fatty acids, ammonia, amines, N-ni- prevents overproduction of mucus, thus maintaining an appro-
troso compounds, phenolic compounds including p-Cresol, priate structure and composition of the gut epithelium (Wrzosek
sulphides, indolic compounds, and hydrogen sulfide. The et al., 2013). Furthermore, dietary fibers can also mechanically
cytotoxic and pro-inflammatory nature of these metabolites stimulate the intestinal epithelium to secrete mucus (McRorie
contributes to the development of chronic diseases, particularly and McKeown, 2017).

708 Cell Host & Microbe 23, June 13, 2018


Cell Host & Microbe

Review

Prolonged lack of dietary fibers damages the mucus barrier obese people have accelerated thymic aging and alteration of
and is associated with increased abundance of mucin-degrad- primary lymphoid tissue architecture (Andersen et al., 2016).
ing bacteria such as Akkermansia muciniphila (Desai et al., Taken together, these observations emphasize the important
2016). Furthermore, when the diet is devoid of dietary fibers role of SCFAs in regulating and maintaining a normal function
some gut bacteria switch their metabolism to use mucin glycans of the innate and adaptive immune system, and although we
by inducing gene expression of mucin-degrading enzymes (Son- have limited the discussion to a few examples, there are several
nenburg et al., 2005). Consistent with this, Western-diet feeding links between fiber intake, immune system, and diseases as
(very low fiber content) of mice increases the penetrability of the illustrated below (Figure 1).
inner mucus layer and lowers growth rate, rendering the mucus
penetrable, and may thus increase the susceptibility to infections The Local Beneficial Effect of Fiber Intake
(Schroeder et al., 2018). Interestingly, low amount of inulin (1%), IBD and CRC
a prebiotic with bifidogenic effect, or Bifidobacterium longum The incidence of IBD is increasing in occidental countries, and
administration prevented mucus defects. Inulin supplementation the Westernization of the diet and lower gut bacterial diversity,
corrected the penetrability of the inner mucus layer, while especially reduction in butyrate-producing bacteria, have been
B. longum supplementation restored the mucus growth rate suggested to contribute to increased IBD prevalence (Ott and
defect, suggesting that those two parameters are independent Schreiber, 2006). In agreement, low fiber intake is correlated
and might be regulated by different factors. Neither 1% inulin with increased incidence of Crohn’s disease (Hou et al., 2011)
nor B. longum administration improved metabolic features of and exacerbates colitis in mice. By investigating the effect of
obese animals. In contrast, high inulin intake (20%) prevented 40 defined diets in mice, Llewellyn et al. demonstrated that
microbiota encroachment, improved gut health, and led to a res- dietary protein and fibers had negative and beneficial effects
olution of the low-grade inflammation associated with improve- on colitis development, respectively. Similarly, mice developed
ment in metabolic parameters of obese mice (Zou et al., 2018). DSS-induced colitis proportional to the amount of fibers in the
Thus, it appears that although low levels of inulin are sufficient diet (Macia et al., 2015). The preventive effect of fibers may be
for restoring local effects in the gut, including protection against due to increased cecal SCFA levels, especially butyrate, which
enteric infections (Desai et al., 2016), higher concentrations are is known to have anti-inflammatory properties, potentially
required for achieving metabolic benefits, suggesting separate through GPR43 (Maslowski et al., 2009).
and dose-dependent mechanisms. However, such high doses IBD can lead to CRC (Beaugerie and Itzkowitz, 2015), which is
of inulin would most likely not be tolerated in humans (Figure 1). the third most common cancer (Johnson et al., 2013). CRC is
An important recent discovery from the Ba €umler lab demon- associated with genetic and environmental factors such as die-
strated that beta-oxidation of butyrate by colonocytes con- tary habits, smoking, and physical activity (Johnson et al.,
sumes oxygen and results in an anaerobic milieu in the gut 2013), and butyrate-producing bacteria are reduced in CRC
(Byndloss et al., 2017). Since butyrate-producing bacteria are patients compared to healthy volunteers (Wang et al., 2012).
very sensitive to oxygen, their abundance is reduced further, Reduced dietary fiber intake is associated with increased inci-
lowering the amount of butyrate production. This feedforward dence of CRC (Aune et al., 2016). Thus, it is not surprising that
loop results in increased luminal oxygen levels, allowing Proteo- diets low in fat and high in fiber-containing grain products, veg-
bacteria such as Escherichia coli and S. enterica serovar Typhi- etables, and fruits have health claims, approved by the FDA, for a
murium to bloom. This novel mechanism provides not only an potential reduction of the incidence of developing some types of
explanation for many of the pathologies associated with a low-fi- cancer. In agreement, a dietary intervention increasing dietary
ber diet, but also a mechanistic understanding for why reduced fibers in African Americans changed the microbiome and
microbial diversity is observed in both humans and mice on a increased butyrogenesis, resulting in reduction of biomarkers
low-fiber diet. of cancer risk (O’Keefe et al., 2015).
The Immune System
A healthy gut microbiota contributes to the maturation and the The Systemic Beneficial Effect of Fiber Intake
development of the immune system (Rescigno, 2014). One Lung COPD and Asthma
such mechanism is through SCFAs, which are known to promote Fiber intake and SCFAs not only act locally but can also affect
generation of colonic regulatory T cells (Tregs) in a GPR43- lung physiology. Patients suffering from severe persistent
dependent manner as well as by inducing histone H3 acetylation asthma consumed more fat and less fiber compared with healthy
(Furusawa et al., 2013; Smith et al., 2013). Accordingly, maternal controls (Berthon et al., 2013). Similarly, a prospective study
high-fiber feeding during pregnancy and lactation modulate showed an inverse relationship between cereal fiber intake and
thymic microenvironment and induced autoimmune regulator the risk of chronic obstructive pulmonary disease (COPD) (Raffa-
(Aire) expression, a factor expressed in the thymus, a primary tellu et al., 2008). In mice, prolonged low-fiber feeding aggra-
lymphoid tissue, which is essential for the maturation of T cells. vated allergic airway disease in mice (Trompette et al., 2014),
The maternal fiber intake increased butyrate levels in the blood which could be corrected by administration of the SCFA propio-
of the offspring and contributed to the enhancement of peripheral nate (Trompette et al., 2014). Thus, it appears that the increase in
and thymic Treg counts of the animals in a GPR41-dependent different airway disease may be coupled to microbial fermenta-
manner (Nakajima et al., 2017). In contrast, high-fat diet induces tion of dietary fibers.
premature thymic involution reflected by the reduced thymocyte Obesity and Diabetes
counts and the increased apoptosis of developing T cell popula- The epidemic of obesity is occurring in both developing and
tions (Yang et al., 2009). These phenotypes may help explain why industrialized countries. Obesity is influenced by several factors

Cell Host & Microbe 23, June 13, 2018 709


Cell Host & Microbe

Review

such as specific dietary and lifestyle behaviors and is associated 1997). Acidification through the production of SCFAs further
with reduced microbial diversity, which may reflect reduced fiber increases mineral solubility and absorption by the colon (Baye
intake. In agreement, a prospective study on 120,877 non-obese et al., 2017; Trinidad et al., 1997). Some of these ions possess
individuals demonstrated that long-term weight gain was anti-microbial action under specific conditions and help in the
inversely correlated with intake of dietary fibers, suggesting their prevention from gut infections. Thus, we hypothesize that the
role in limiting long-term body weight gain (Mozaffarian et al., capacity of different dietary fibers to bind ions in the gut may
2011). Similarly, high fiber intake is associated with increased serve to establish important local reservoirs. Accordingly, zinc
gut microbial diversity and lower long-term weight gain (Menni has been shown to promote the metabolic activity of gut micro-
et al., 2017). A recent intervention study with oligofructose- biota of weaned piglets (Højberg et al., 2005), resulting in
enriched inulin for 16 weeks in overweight and obese children improved metabolic health parameters. In parallel, a recent
reduced their fat mass, suggesting that increased intake of study in chickens demonstrated that zinc-deficient diet results
fermentable fibers may have beneficial effects on obesity (Nico- in lower gut microbiota diversity associated with reduced
lucci et al., 2017). SCFA production (Reed et al., 2015). These observations sug-
Obesity is associated with type 2 diabetes (T2D). In contrast to gest that dietary fibers could participate in preserving a healthy
obesity, T2D is associated with reduced abundance of fiber-de- intestinal ecology by providing micronutrients to bacteria and
grading bacteria (Karlsson et al., 2013; Qin et al., 2012). Accord- the host in the distal gut. However, some studies suggested
ingly, diets with high glucose index (high in digestible starch and that mineral binding to dietary fibers may lower their availability
low in fiber) are associated with an increased diabetes risk (Sluijs for the host, which may lead to mineral deficiency (Baye et al.,
et al., 2012). Administration of soluble fibers, such as oligofruc- 2017). It is thus important to determine the mineral binding
tose and long-chain inulin, corrected gut dysbiosis, reduced capacity and effects on homeostasis for different fibers.
body weight gain and low-grade inflammation, and improved In addition to regulating bioavailability to nutrients as sug-
glucose metabolism, which was at least attributed to decreased gested above, fibers may also form platforms for bringing bacte-
intestinal permeability and endotoxemia (Cani et al., 2007, 2008). ria and biomolecules into close proximity. For example, conju-
Furthermore, providing healthy human volunteers with barley gated bile acids in the small intestine can bind to fibers before
kernel-based bread (BKB), rich in b-glucans, improved glucose they are deconjugated by Bacteroides and Lacotobacillus
metabolism (Nilsson et al., 2015). Thus, it is tempting to specu- species (Kahlon and Woodruff, 2003). Deconjugation is a prereq-
late that whereas the reduced diversity in obesity is caused by uisite for further biotransformation to secondary bile acids by low
reduced fiber intake, lack of fiber-degrading and butyrate-pro- abundant bacteria such as Clostridium scindens (Reddy et al.,
ducing bacteria may predispose to T2D. 1992). We thus suggest that binding of both bile acids and spe-
cific bacteria to the same fiber, which serves as a platform, can
SCFA-Independent Effect of Dietary Fibers explain how low abundant bacteria can serve as efficient trans-
Thus far, this review has focused mainly on the fermentation of formers of biomolecules and thus increase production of
fibers to SCFAs, but it is important to mention that microbial secondary bile acids. Secondary bile acids, such as deoxycholic
metabolism of fibers also has additional effects. Ferulic acid acids, have a number of physiologically important effects and
(FA) is a phenolic compound that can be found in plant cell have been positively associated with both CRC (Bernstein
wall and serves to enhance its rigidity and strength. Microbial et al., 2005) and improved metabolism (Li and Chiang, 2015)
metabolism of dietary fibers, for example those found in cereal (Figure 2). This platform concept can perhaps be extended to
bran, leads to the release of FA by bacteria such as additional bacteria and biomolecules.
L. fermentum NCIMB 5221 harboring the FA esterase gene
(Tomaro-Duchesneau et al., 2012). In the digestive tract, FA One Size Does Not Fit All: Fiber Quantity and
can either act locally to modulate gut physiology or be trans- Personalization
ported in its free form into the bloodstream to influence systemic The role of dietary fibers in preventing and alleviating chronic
health. FA has antioxidant and anti-inflammatory properties and inflammatory diseases in humans has been widely studied
could be considered as a potential therapeutic treatment for during the past years, although findings from intervention trials
various chronic pathologies such as neurodegeneration, obesity, are often inconsistent (Buyken et al., 2014). Studies performed
diabetes, and cancer. FA administration stimulates neurogene- on animal models usually have more dramatic effects, but also
sis in corticosterone-treated mice and was shown to prevent use higher amount of fibers compared to levels used in human
Ab-related toxicity in Alzheimer disease models (Mori et al., clinical trials, especially for prebiotics, whose dose is often
2013). In a model of ulcerative colitis, FA treatment showed 40-fold higher on a body weight basis (Schaafsma and Slavin,
anti-inflammatory properties reflected by the reduction of pro-in- 2014). In fact, the dose of dietary fiber used in animal studies
flammatory cytokine and the upregulation of IL-10 (Sadar et al., more closely resembles the estimated amount of fiber
2016). In addition, FA can prevent development of diet-induced consumed by our ancestors before the advent of agriculture
obesity (de Melo et al., 2017) and has anti-diabetic effects: dia- (>100 g/day) (Eaton et al., 1997). Therefore, even fiber doses
betic rats treated with this compound normalized glucose and that meet the dietary recommendations of today (30 g/day)
serum insulin levels (Narasimhan et al., 2015). (Jones, 2014) are still far below the fiber amounts ingested
Dietary fibers can also bind different micro- and macronutri- when the symbiosis between us and the microbes was formed
ents including ions such as copper, calcium, and zinc and trans- (Deehan and Walter, 2016). Non-industrialized human popula-
port them to the distal gut, where they are released when the tions that consume fiber amounts of more than 50 g/day, such
fiber is metabolized by the colonic bacteria (Bergman et al., as rural South Africans and rural Ugandans, are known to be

710 Cell Host & Microbe 23, June 13, 2018


Cell Host & Microbe

Review
Figure 2. SCFA-Independent Effect of Dietary
Fibers in Colon
Dietary fibers bind conjugated primary bile acids
BSH 7α-dehydroxylation Gut bacteria (BAs) and may serve as a platform for gut bacteria
that possess the bile salt hydrolase (Bsh), leading to
the production of non-conjugated BAs. These can
also bind to dietary fibers and be further metabolized
by specific bacteria with 7-alpha dehydroxylation
activity, thus generating secondary BAs. The fact
that dietary fibers can bind secondary BAs suggests
that they may play a role in regulating BA levels
within the gut. This structural interaction may
modulate host physiology either by preventing the
accumulation of toxic BAs that can lead to the
development of polyps and colorectal cancer (CRC)
or by increasing the disposal of BAs that can acti-
Increased colonic vate TGR5 to increase glucagon-like peptide 1
absorption (GLP-1) secretion. In addition, bacterial degradation
of dietary fibers leads to the release of minerals and
phenolic compounds, which can be absorbed by the
distal gut.

Colon polyps can lead to undesirable side effects, such


as flatulence, bloating, stomachaches,
diarrhea, and constipation (Grabitske
and Slavin, 2009), and may thus be nega-
Increased tive for individuals with irritable bowel syn-
GLP-1 colonic excretion Polyp formation
drome. In addition, a few recent studies
using animal models suggest that fiber
intake and/or their derived metabolites
may have a negative impact on host
Conjugated Phenolic compounds health in specific conditions such as coli-
primary bile acids tis (Miles et al., 2017) and/or CRC (Bel-
Minerals and ions cheva et al., 2014). Although the over-
Unconjugated whelming data suggest beneficial effects
primary bile acids Carbohydrate of fibers, there may be instances in which
binding protein caution in making generalizable recom-
Secondary mendations is warranted. It should be
bile acids TGR5 noted that tolerance to fiber is dependent
on the individual and often improves over
time as the gastrointestinal tract and mi-
crobiota adapt to higher doses of dietary
fiber (Mego et al., 2017). It is therefore
largely free from chronic inflammatory diseases. Most impor- realistic to have the diet of modern humans enriched with fiber
tantly, moving African Americans to an ‘‘African-style’’ diet with beyond 50 g daily, especially with sufficient acclimatization and
55 g fiber reversed risk markers of CRC within only 2 weeks if slower fermenting polysaccharides, such as RS, arabinoxy-
(O’Keefe et al., 2015). Therefore, it is plausible that dietary fiber lan, acacia gum, and resistant maltodextrin, are consumed
supplementation below these doses, which are used in virtually since these fiber types are better tolerated at higher doses
all human intervention trials today, are too low and physiologi- than faster fermenting oligosaccharides like oligofructose or
cally irrelevant. Recent suggestions postulate daily fiber galactooligosaccharides (Deehan and Walter, 2016). For future
amounts of greater than 50 g to achieve health benefits linked applications, one could use specific fiber types in a personal-
to fiber (Deehan et al., 2017; O’Keefe, 2018), and accordingly, ized fashion (matched with microbiota profile) to reduce the
human interventions with greater than 50 g/day of fiber observed severity of side effects while enhancing the physiologic benefit
significant improvements in the assessed health markers (Jen- to the host.
kins et al., 2001; O’Keefe et al., 2015; Pedersen et al., 2013; Inconsistencies in human intervention trials could also result
Zhao et al., 2018). However, high fiber amounts will be chal- from the response of the human gut microbiota to dietary fiber
lenging to achieve with regular food items but could be achieved being highly individualized (Davis et al., 2011; Martı́nez et al.,
by systematically supplementing the food supply with dietary 2010). Missing keystone species in individuals, the absence of
fiber sources (Deehan and Walter, 2016). functional ‘‘guilds’’ able to assess a fiber, and/or the absence
There are concerns, however, that modern humans may of strains that can utilize specific fiber sources could explain
have problems tolerating such high doses of fiber as its intake why some studies show segregation in response profiles of

Cell Host & Microbe 23, June 13, 2018 711


Cell Host & Microbe

Review

patients during dietary intervention (Deehan et al., 2017). Match- of Synbiotics Solutions, a developer of synbiotic products. F.B. is founder and
shareholder of Metabogen AB.
ing of fibers to the microbiota may produce more beneficial ef-
fects on the host, especially if the subject’s lack of keystone spe-
REFERENCES
cies leads to a non-response profile. This was exemplified in a
recent study showing that supplementation of BKB did not result
Andersen, C.J., Murphy, K.E., and Fernandez, M.L. (2016). Impact of obesity
in metabolic improvement in all healthy subjects. Individuals who and metabolic syndrome on immunity. Adv. Nutr. 7, 66–75.
exhibited improved metabolic profile after 3 days of BKB con-
Aune, D., Keum, N., Giovannucci, E., Fadnes, L.T., Boffetta, P., Greenwood,
sumption had a higher Prevotella/Bacteriodes ratio with an D.C., Tonstad, S., Vatten, L.J., Riboli, E., and Norat, T. (2016). Whole grain con-
enrichment of Prevotella copri, reflecting the ability to degrade sumption and risk of cardiovascular disease, cancer, and all cause and cause
complex carbohydrates (Kovatcheva-Datchary et al., 2015). specific mortality: systematic review and dose-response meta-analysis of pro-
spective studies. BMJ 353, i2716.
Interestingly, the capacity for using fibers varies between Prevo-
tella- versus Bacteroides-dominated gut microbiota. Prevotella- Ba€ckhed, F., Ding, H., Wang, T., Hooper, L.V., Koh, G.Y., Nagy, A., Semenko-
vich, C.F., and Gordon, J.I. (2004). The gut microbiota as an environmental
dominated bacteria produce higher levels and different propor- factor that regulates fat storage. Proc. Natl. Acad. Sci. USA 101, 15718–15723.
tions of SCFAs compared to Bacteroides-dominated microbiota
(Chen et al., 2017). In parallel, a very elegant study performed on Baye, K., Guyot, J.P., and Mouquet-Rivier, C. (2017). The unresolved role of
dietary fibers on mineral absorption. Crit. Rev. Food Sci. Nutr. 57, 949–957.
800 individuals, representative of an adult non-prediabetic
population, showed that post-prandial glucose response Beaugerie, L., and Itzkowitz, S.H. (2015). Cancers complicating inflammatory
bowel disease. N. Engl. J. Med. 373, 195.
(PPGR) was highly variable between individuals who consumed
the same standardized meal. The PPGR correlated with some Belcheva, A., Irrazabal, T., Robertson, S.J., Streutker, C., Maughan, H., Ru-
bino, S., Moriyama, E.H., Copeland, J.K., Surendra, A., Kumar, S., et al.
microbiota features, highlighting the importance of the diet-mi-
(2014). Gut microbial metabolism drives transformation of MSH2-deficient
crobiota interaction and its impact on host metabolism. An colon epithelial cells. Cell 158, 288–299.
algorithm using clinical and microbiota profiles as input demon-
Bergman, C.J., Gualberto, D.G., and Weber, C.W. (1997). Mineral binding ca-
strated that the PPGR of each individual can be predicted and pacity of dephytinized insoluble fiber from extruded wheat, oat and rice brans.
that a personalized dietary intervention based on their predictor Plant Foods Hum. Nutr. 51, 295–310.
can lead to an improvement in glucose metabolism including Bernstein, H., Bernstein, C., Payne, C.M., Dvorakova, K., and Garewal, H.
lower PPGR (Zeevi et al., 2015). (2005). Bile acids as carcinogens in human gastrointestinal cancers. Mutat.
Res. 589, 47–65.
Concluding Remarks Berthon, B.S., Macdonald-Wicks, L.K., Gibson, P.G., and Wood, L.G. (2013).
In conclusion, dietary fibers can be considered key ancestral Investigation of the association between dietary intake, disease severity and
airway inflammation in asthma. Respirology 18, 447–454.
compounds that preserve gut ecology, especially regulating
macronutrients and host physiology. Screening novel fibers, Bindels, L.B., Delzenne, N.M., Cani, P.D., and Walter, J. (2015). Towards a
both extracted and purified from food as well as those selectively more comprehensive concept for prebiotics. Nat. Rev. Gastroenterol. Hepatol.
12, 303–310.
modified or synthesized, for their potential as the next-genera-
tion prebiotics, and defining efficient strategies to reintroduce Buyken, A.E., Goletzke, J., Joslowski, G., Felbick, A., Cheng, G., Herder, C.,
and Brand-Miller, J.C. (2014). Association between carbohydrate quality and
high amount of fibers aiming at replenishing the gut microbiome inflammatory markers: systematic review of observational and interventional
with essential missing microbes, will be the next challenge to studies. Am. J. Clin. Nutr. 99, 813–833.
significantly impact gut microbiota-associated human diseases.
Byndloss, M.X., Olsan, E.E., Rivera-Chávez, F., Tiffany, C.R., Cevallos, S.A.,
Finally, a better understanding of diet-microbiota interactions Lokken, K.L., Torres, T.P., Byndloss, A.J., Faber, F., Gao, Y., et al. (2017).
will help to develop a personalized nutrition approach that would Microbiota-activated PPAR-g signaling inhibits dysbiotic Enterobacteriaceae
expansion. Science 357, 570–575.
target and reduce more efficiently the incidence of chronic in-
flammatory diseases. Cani, P.D., Neyrinck, A.M., Fava, F., Knauf, C., Burcelin, R.G., Tuohy, K.M.,
Gibson, G.R., and Delzenne, N.M. (2007). Selective increases of bifidobac-
teria in gut microflora improve high-fat-diet-induced diabetes in mice
ACKNOWLEDGMENTS
through a mechanism associated with endotoxaemia. Diabetologia 50,
2374–2383.
We are grateful to Anna Hallén for producing the figures. This study was sup-
ported by the Novo Nordisk Foundation, the Swedish Research Council, the Cani, P.D., Bibiloni, R., Knauf, C., Waget, A., Neyrinck, A.M., Delzenne, N.M.,
Swedish Diabetes Foundation, the Swedish Heart Lung Foundation, Göran and Burcelin, R. (2008). Changes in gut microbiota control metabolic endotox-
Gustafsson’s Foundation, Knut and Alice Wallenberg Foundation, the FP7- emia-induced inflammation in high-fat diet-induced obesity and diabetes in
sponsored program METACARDIS, JPI (healthy diet for a healthy life), the mice. Diabetes 57, 1470–1481.
regional agreement on medical training and clinical research (ALF) between
Chen, T., Long, W., Zhang, C., Liu, S., Zhao, L., and Hamaker, B.R. (2017). Fi-
Region Va €stra Götaland and Sahlgrenska University Hospital, and a grant
ber-utilizing capacity varies in Prevotella- versus Bacteroides-dominated gut
from a Transatlantic Networks of Excellence Award from the Leducq Founda- microbiota. Sci. Rep. 7, 2594.
tion. F.B. is Torsten Söderberg Professor in Medicine and recipient of an ERC
Consolidator Grant (European Research Council, Consolidator grant 615362 - Clemente, J.C., Pehrsson, E.C., Blaser, M.J., Sandhu, K., Gao, Z., Wang, B.,
METABASE). J.W. is a Campus Alberta Innovation Program (CAIP) chair for Magris, M., Hidalgo, G., Contreras, M., Noya-Alarcón, Ó., et al. (2015). The
Nutrition, Microbes, and Gastrointestinal Health and recipient of grants from microbiome of uncontacted Amerindians. Sci. Adv. 1, e1500183.
the Canadian Institutes of Health Research (CIHR) and JPI (healthy diet for a
healthy life). Cummings, J.H., and Macfarlane, G.T. (1991). The control and consequences
of bacterial fermentation in the human colon. J. Appl. Bacteriol. 70, 443–459.
DECLARATION OF INTERESTS David, L.A., Maurice, C.F., Carmody, R.N., Gootenberg, D.B., Button, J.E.,
Wolfe, B.E., Ling, A.V., Devlin, A.S., Varma, Y., Fischbach, M.A., et al.
J.W. has received research funding from industry sources involved in the (2014). Diet rapidly and reproducibly alters the human gut microbiome. Nature
manufacture and marketing of prebiotics and dietary fibers, and is a co-owner 505, 559–563.

712 Cell Host & Microbe 23, June 13, 2018


Cell Host & Microbe

Review
Davis, L.M., Martı́nez, I., Walter, J., Goin, C., and Hutkins, R.W. (2011). Bar- Jones, J.M. (2014). CODEX-aligned dietary fiber definitions help to bridge the
coded pyrosequencing reveals that consumption of galactooligosaccharides ‘fiber gap’. Nutr. J. 13, 34.
results in a highly specific bifidogenic response in humans. PLoS One 6,
e25200. Kahlon, T.S., and Woodruff, C.L. (2003). In vitro binding of bile acids by rice
bran, oat bran, barley and b-glucan enriched barley. Cereal Chem. 80,
De Filippo, C., Cavalieri, D., Di Paola, M., Ramazzotti, M., Poullet, J.B., Mas- 260–263.
sart, S., Collini, S., Pieraccini, G., and Lionetti, P. (2010). Impact of diet in
shaping gut microbiota revealed by a comparative study in children from Karlsson, F.H., Tremaroli, V., Nookaew, I., Bergström, G., Behre, C.J., Fager-
Europe and rural Africa. Proc. Natl. Acad. Sci. USA 107, 14691–14696. €ckhed, F. (2013). Gut metagenome in European
berg, B., Nielsen, J., and Ba
women with normal, impaired and diabetic glucose control. Nature
De Filippo, C., Di Paola, M., Ramazzotti, M., Albanese, D., Pieraccini, G., 498, 99–103.
Banci, E., Miglietta, F., Cavalieri, D., and Lionetti, P. (2017). Diet, environments,
and gut microbiota. a preliminary investigation in children living in rural and Koh, A., De Vadder, F., Kovatcheva-Datchary, P., and Ba €ckhed, F. (2016).
urban Burkina Faso and Italy. Front. Microbiol. 8, 1979. From dietary fiber to host physiology: short-chain fatty acids as key bacterial
metabolites. Cell 165, 1332–1345.
de Melo, T.S., Lima, P.R., Carvalho, K.M., Fontenele, T.M., Solon, F.R., Tomé,
A.R., de Lemos, T.L., da Cruz Fonseca, S.G., Santos, F.A., Rao, V.S., and de Kovatcheva-Datchary, P., Nilsson, A., Akrami, R., Lee, Y.S., De Vadder, F., Ar-
Queiroz, M.G. (2017). Ferulic acid lowers body weight and visceral fat accumu- €ckhed, F. (2015). Dietary fiber-
ora, T., Hallen, A., Martens, E., Björck, I., and Ba
lation via modulation of enzymatic, hormonal and inflammatory changes in a induced improvement in glucose metabolism is associated with increased
mouse model of high-fat diet-induced obesity. Braz. J. Med. Biol. Res. abundance of Prevotella. Cell Metab. 22, 971–982.
50, e5630.
Li, T., and Chiang, J.Y. (2015). Bile acids as metabolic regulators. Curr. Opin.
Deehan, E.C., and Walter, J. (2016). The fiber gap and the disappearing gut mi- Gastroenterol. 31, 159–165.
crobiome: implications for human nutrition. Trends Endocrinol. Metab. 27,
239–242. Logan, A.C., Jacka, F.N., and Prescott, S.L. (2016). Immune-microbiota inter-
actions: dysbiosis as a global health issue. Curr. Allergy Asthma Rep. 16, 13.
Deehan, E.C., Duar, R.M., Armet, A.M., Perez-Muñoz, M.E., Jin, M., and Wal-
ter, J. (2017). Modulation of the gastrointestinal microbiome with nondigestible Lovegrove, A., Edwards, C.H., De Noni, I., Patel, H., El, S.N., Grassby, T.,
fermentable carbohydrates to improve human health. Microbiol. Spectr. 5, Zielke, C., Ulmius, M., Nilsson, L., Butterworth, P.J., et al. (2017). Role of poly-
https://doi.org/10.1128/microbiolspec.BAD-0019-2017. saccharides in food, digestion, and health. Crit. Rev. Food Sci. Nutr. 57,
237–253.
Desai, M.S., Seekatz, A.M., Koropatkin, N.M., Kamada, N., Hickey, C.A., Wol-
ter, M., Pudlo, N.A., Kitamoto, S., Terrapon, N., Muller, A., et al. (2016). A
Macia, L., Tan, J., Vieira, A.T., Leach, K., Stanley, D., Luong, S., Maruya, M.,
dietary fiber-deprived gut microbiota degrades the colonic mucus barrier
Ian McKenzie, C., Hijikata, A., Wong, C., et al. (2015). Metabolite-sensing re-
and enhances pathogen susceptibility. Cell 167, 1339–1353.e21.
ceptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis
through regulation of the inflammasome. Nat. Commun. 6, 6734.
Duncan, S.H., Belenguer, A., Holtrop, G., Johnstone, A.M., Flint, H.J., and Lob-
ley, G.E. (2007). Reduced dietary intake of carbohydrates by obese subjects € sa
€nen,
Mardinoglu, A., Wu, H., Bjornson, E., Zhang, C., Hakkarainen, A., Ra
results in decreased concentrations of butyrate and butyrate-producing
€inen, K.H., et al. (2018).
S.M., Lee, S., Mancina, R.M., Bergentall, M., Pietila
bacteria in feces. Appl. Environ. Microbiol. 73, 1073–1078.
An integrated understanding of the rapid metabolic benefits of a carbohy-
drate-restricted diet on hepatic steatosis in humans. Cell Metab. 27,
Eaton, S.B., Eaton, S.B., 3rd, and Konner, M.J. (1997). Paleolithic nutrition re-
559–571.e5.
visited: a twelve-year retrospective on its nature and implications. Eur. J. Clin.
Nutr. 51, 207–216.
Martı́nez, I., Kim, J., Duffy, P.R., Schlegel, V.L., and Walter, J. (2010). Resistant
Flint, H.J., Scott, K.P., Duncan, S.H., Louis, P., and Forano, E. (2012). Microbial starches types 2 and 4 have differential effects on the composition of the fecal
degradation of complex carbohydrates in the gut. Gut Microbes 3, 289–306. microbiota in human subjects. PLoS One 5, e15046.

Furusawa, Y., Obata, Y., Fukuda, S., Endo, T.A., Nakato, G., Takahashi, D., Martı́nez, I., Stegen, J.C., Maldonado-Gómez, M.X., Eren, A.M., Siba, P.M.,
Nakanishi, Y., Uetake, C., Kato, K., Kato, T., et al. (2013). Commensal Greenhill, A.R., and Walter, J. (2015). The gut microbiota of rural papua new
microbe-derived butyrate induces the differentiation of colonic regulatory guineans: composition, diversity patterns, and ecological processes. Cell
T cells. Nature 504, 446–450. Rep. 11, 527–538.

Grabitske, H.A., and Slavin, J.L. (2009). Gastrointestinal effects of low-digest- Maslowski, K.M., Vieira, A.T., Ng, A., Kranich, J., Sierro, F., Yu, D., Schilter,
ible carbohydrates. Crit. Rev. Food Sci. Nutr. 49, 327–360. H.C., Rolph, M.S., Mackay, F., Artis, D., et al. (2009). Regulation of inflamma-
tory responses by gut microbiota and chemoattractant receptor GPR43.
Hand, T.W., Vujkovic-Cvijin, I., Ridaura, V.K., and Belkaid, Y. (2016). Linking Nature 461, 1282–1286.
the microbiota, chronic disease, and the immune system. Trends Endocrinol.
Metab. 27, 831–843. McRorie, J.W., Jr., and McKeown, N.M. (2017). Understanding the physics of
functional fibers in the gastrointestinal tract: an evidence-based approach to
Højberg, O., Canibe, N., Poulsen, H.D., Hedemann, M.S., and Jensen, B.B. resolving enduring misconceptions about insoluble and soluble fiber.
(2005). Influence of dietary zinc oxide and copper sulfate on the gastrointes- J. Acad. Nutr. Diet. 117, 251–264.
tinal ecosystem in newly weaned piglets. Appl. Environ. Microbiol. 71,
2267–2277. Mego, M., Accarino, A., Tzortzis, G., Vulevic, J., Gibson, G., Guarner, F., and
Azpiroz, F. (2017). Colonic gas homeostasis: Mechanisms of adaptation
Hou, J.K., Abraham, B., and El-Serag, H. (2011). Dietary intake and risk of following HOST-G904 galactooligosaccharide use in humans. Neurogastroen-
developing inflammatory bowel disease: a systematic review of the literature. terol. Motil. 29, https://doi.org/10.1111/nmo.13080.
Am. J. Gastroenterol. 106, 563–573.
Menni, C., Jackson, M.A., Pallister, T., Steves, C.J., Spector, T.D., and Valdes,
Jenkins, D.J., Kendall, C.W., Popovich, D.G., Vidgen, E., Mehling, C.C., Vuk- A.M. (2017). Gut microbiome diversity and high-fibre intake are related to lower
san, V., Ransom, T.P., Rao, A.V., Rosenberg-Zand, R., Tariq, N., et al. long-term weight gain. Int. J. Obes. 41, 1099–1105.
(2001). Effect of a very-high-fiber vegetable, fruit, and nut diet on serum lipids
and colonic function. Metabolism 50, 494–503. Miles, J.P., Zou, J., Kumar, M.V., Pellizzon, M., Ulman, E., Ricci, M., Gewirtz,
A.T., and Chassaing, B. (2017). Supplementation of low- and high-fat diets with
Johansson, M.E., Phillipson, M., Petersson, J., Velcich, A., Holm, L., and Hans- fermentable fiber exacerbates severity of DSS-induced acute colitis. Inflamm.
son, G.C. (2008). The inner of the two Muc2 mucin-dependent mucus layers in Bowel Dis. 23, 1133–1143.
colon is devoid of bacteria. Proc. Natl. Acad. Sci. USA 105, 15064–15069.
Mori, T., Koyama, N., Guillot-Sestier, M.V., Tan, J., and Town, T. (2013).
Johnson, C.M., Wei, C., Ensor, J.E., Smolenski, D.J., Amos, C.I., Levin, B., and Ferulic acid is a nutraceutical b-secretase modulator that improves behavioral
Berry, D.A. (2013). Meta-analyses of colorectal cancer risk factors. Cancer impairment and alzheimer-like pathology in transgenic mice. PLoS One 8,
Causes Control 24, 1207–1222. e55774.

Cell Host & Microbe 23, June 13, 2018 713


Cell Host & Microbe

Review
Mozaffarian, D., Hao, T., Rimm, E.B., Willett, W.C., and Hu, F.B. (2011). Sluijs, I., Forouhi, N.G., Beulens, J.W., van der Schouw, Y.T., Agnoli, C., Ar-
Changes in diet and lifestyle and long-term weight gain in women and men. riola, L., Balkau, B., Barricarte, A., Boeing, H., Bueno-de-Mesquita, H.B.,
N. Engl. J. Med. 364, 2392–2404. et al.; InterAct Consortium (2012). The amount and type of dairy product intake
and incident type 2 diabetes: results from the EPIC-InterAct Study. Am. J. Clin.
Nakajima, A., Kaga, N., Nakanishi, Y., Ohno, H., Miyamoto, J., Kimura, I., Hori, Nutr. 96, 382–390.
S., Sasaki, T., Hiramatsu, K., Okumura, K., et al. (2017). Maternal high fiber diet
during pregnancy and lactation influences regulatory T cell differentiation in Smith, P.M., Howitt, M.R., Panikov, N., Michaud, M., Gallini, C.A., Bohlooly-Y,
offspring in mice. J. Immunol. 199, 3516–3524. M., Glickman, J.N., and Garrett, W.S. (2013). The microbial metabolites, short-
chain fatty acids, regulate colonic Treg cell homeostasis. Science 341,
Narasimhan, A., Chinnaiyan, M., and Karundevi, B. (2015). Ferulic acid exerts 569–573.
its antidiabetic effect by modulating insulin-signalling molecules in the liver of
high-fat diet and fructose-induced type-2 diabetic adult male rat. Appl. Phys- Smits, S.A., Leach, J., Sonnenburg, E.D., Gonzalez, C.G., Lichtman, J.S.,
iol. Nutr. Metab. 40, 769–781. Reid, G., Knight, R., Manjurano, A., Changalucha, J., Elias, J.E., et al. (2017).
Seasonal cycling in the gut microbiome of the Hadza hunter-gatherers of
Nicolucci, A.C., Hume, M.P., Martı́nez, I., Mayengbam, S., Walter, J., and Re- Tanzania. Science 357, 802–806.
imer, R.A. (2017). Prebiotics reduce body fat and alter intestinal microbiota in
children who are overweight or with obesity. Gastroenterology 153, 711–722. Sommer, F., Ståhlman, M., Ilkayeva, O., Arnemo, J.M., Kindberg, J., Josefs-
son, J., Newgard, C.B., Fröbert, O., and Ba €ckhed, F. (2016). The gut
Nilsson, A.C., Johansson-Boll, E.V., and Björck, I.M. (2015). Increased gut hor- microbiota modulates energy metabolism in the hibernating brown bear Ursus
mones and insulin sensitivity index following a 3-d intervention with a barley arctos. Cell Rep. 14, 1655–1661.
kernel-based product: a randomised cross-over study in healthy middle-
aged subjects. Br. J. Nutr. 114, 899–907. Sonnenburg, E.D., and Sonnenburg, J.L. (2014). Starving our microbial self:
the deleterious consequences of a diet deficient in microbiota-accessible
O’Keefe, S.J.D. (2018). The need to reassess dietary fiber requirements in carbohydrates. Cell Metab. 20, 779–786.
healthy and critically ill patients. Gastroenterol. Clin. North Am. 47, 219–229.
Sonnenburg, J.L., Xu, J., Leip, D.D., Chen, C.H., Westover, B.P., Weatherford,
O’Keefe, S.J., Li, J.V., Lahti, L., Ou, J., Carbonero, F., Mohammed, K., Posma, J., Buhler, J.D., and Gordon, J.I. (2005). Glycan foraging in vivo by an intestine-
J.M., Kinross, J., Wahl, E., Ruder, E., et al. (2015). Fat, fibre and cancer risk in adapted bacterial symbiont. Science 307, 1955–1959.
African Americans and rural Africans. Nat. Commun. 6, 6342.
Sonnenburg, E.D., Smits, S.A., Tikhonov, M., Higginbottom, S.K., Wingreen,
Ott, S.J., and Schreiber, S. (2006). Reduced microbial diversity in inflammatory N.S., and Sonnenburg, J.L. (2016). Diet-induced extinctions in the gut micro-
bowel diseases. Gut 55, 1207. biota compound over generations. Nature 529, 212–215.

Pedersen, C., Lefevre, S., Peters, V., Patterson, M., Ghatei, M.A., Morgan, Stephen, A.M., Champ, M.M., Cloran, S.J., Fleith, M., van Lieshout, L., Mej-
L.M., and Frost, G.S. (2013). Gut hormone release and appetite regulation in born, H., and Burley, V.J. (2017). Dietary fibre in Europe: current state of
healthy non-obese participants following oligofructose intake. A dose-escala- knowledge on definitions, sources, recommendations, intakes and relation-
tion study. Appetite 66, 44–53. ships to health. Nutr. Res. Rev. 30, 149–190.

Qin, J., Li, Y., Cai, Z., Li, S., Zhu, J., Zhang, F., Liang, S., Zhang, W., Guan, Y., Tomaro-Duchesneau, C., Saha, S., Malhotra, M., Coussa-Charley, M., Ka-
Shen, D., et al. (2012). A metagenome-wide association study of gut micro- houli, I., Jones, M.L., Labbé, A., and Prakash, S. (2012). Probiotic ferulic
biota in type 2 diabetes. Nature 490, 55–60. acid esterase active Lactobacillus fermentum NCIMB 5221 APA microcap-
sules for oral delivery: preparation and in vitro characterization. Pharmaceuti-
Raffatellu, M., Santos, R.L., Verhoeven, D.E., George, M.D., Wilson, R.P., cals (Basel) 5, 236–248.
Winter, S.E., Godinez, I., Sankaran, S., Paixao, T.A., Gordon, M.A., et al.
(2008). Simian immunodeficiency virus-induced mucosal interleukin-17 defi- Trinidad, T.P., Wolever, T.M., and Thompson, L.U. (1997). Effect of short chain
ciency promotes Salmonella dissemination from the gut. Nat. Med. 14, fatty acids on calcium absorption in humans. Adv. Exp. Med. Biol. 427,
421–428. 183–189.

Reddy, B.S., Engle, A., Simi, B., and Goldman, M. (1992). Effect of dietary fiber Trompette, A., Gollwitzer, E.S., Yadava, K., Sichelstiel, A.K., Sprenger, N.,
on colonic bacterial enzymes and bile acids in relation to colon cancer. Gastro- Ngom-Bru, C., Blanchard, C., Junt, T., Nicod, L.P., Harris, N.L., and Marsland,
enterology 102, 1475–1482. B.J. (2014). Gut microbiota metabolism of dietary fiber influences allergic
airway disease and hematopoiesis. Nat. Med. 20, 159–166.
Reed, S., Neuman, H., Moscovich, S., Glahn, R.P., Koren, O., and Tako, E.
(2015). Chronic zinc deficiency alters chick gut microbiota composition and Walker, A.W., Ince, J., Duncan, S.H., Webster, L.M., Holtrop, G., Ze, X., Brown,
function. Nutrients 7, 9768–9784. D., Stares, M.D., Scott, P., Bergerat, A., et al. (2011). Dominant and diet-
responsive groups of bacteria within the human colonic microbiota. ISME J.
Rescigno, M. (2014). Intestinal microbiota and its effects on the immune sys- 5, 220–230.
tem. Cell. Microbiol. 16, 1004–1013.
Walter, J., Britton, R.A., and Roos, S. (2011). Host-microbial symbiosis in the
Sadar, S.S., Vyawahare, N.S., and Bodhankar, S.L. (2016). Ferulic acid amelio- vertebrate gastrointestinal tract and the Lactobacillus reuteri paradigm.
rates TNBS-induced ulcerative colitis through modulation of cytokines, oxida- Proc. Natl. Acad. Sci. USA 108 (Suppl 1 ), 4645–4652.
tive stress, iNOs, COX-2, and apoptosis in laboratory rats. EXCLI J. 15,
482–499. Wang, T., Cai, G., Qiu, Y., Fei, N., Zhang, M., Pang, X., Jia, W., Cai, S., and
Zhao, L. (2012). Structural segregation of gut microbiota between colorectal
Schaafsma, G., and Slavin, J.L. (2014). Significance of inulin fructans in the hu- cancer patients and healthy volunteers. ISME J. 6, 320–329.
man diet. Compr. Rev. Food Sci. Food Saf. 14, 37–47.
Windey, K., De Preter, V., and Verbeke, K. (2012). Relevance of protein fermen-
Schnorr, S.L., Candela, M., Rampelli, S., Centanni, M., Consolandi, C., Basa- tation to gut health. Mol. Nutr. Food Res. 56, 184–196.
glia, G., Turroni, S., Biagi, E., Peano, C., Severgnini, M., et al. (2014). Gut
microbiome of the Hadza hunter-gatherers. Nat. Commun. 5, 3654. Wrzosek, L., Miquel, S., Noordine, M.L., Bouet, S., Joncquel Chevalier-Curt,
M., Robert, V., Philippe, C., Bridonneau, C., Cherbuy, C., Robbe-Masselot,
Schroeder, B.O., and Ba €ckhed, F. (2016). Signals from the gut microbiota to C., et al. (2013). Bacteroides thetaiotaomicron and Faecalibacterium praus-
distant organs in physiology and disease. Nat. Med. 22, 1079–1089. nitzii influence the production of mucus glycans and the development of
goblet cells in the colonic epithelium of a gnotobiotic model rodent. BMC
Schroeder, B.O., Birchenough, G.M.H., Ståhlman, M., Arike, L., Johansson, Biol. 11, 61.
€ckhed, F. (2018). Bifidobacteria or fiber protects
M.E.V., Hansson, G.C., and Ba
against diet-induced microbiota-mediated colonic mucus deterioration. Cell Wu, G.D., Chen, J., Hoffmann, C., Bittinger, K., Chen, Y.Y., Keilbaugh, S.A.,
Host Microbe 23, 27–40.e7. Bewtra, M., Knights, D., Walters, W.A., Knight, R., et al. (2011). Linking long-
term dietary patterns with gut microbial enterotypes. Science 334, 105–108.
Schwarzer, M., Makki, K., Storelli, G., Machuca-Gayet, I., Srutkova, D., Her-
manova, P., Martino, M.E., Balmand, S., Hudcovic, T., Heddi, A., et al. Yang, H., Youm, Y.H., Vandanmagsar, B., Rood, J., Kumar, K.G., Butler, A.A.,
(2016). Lactobacillus plantarum strain maintains growth of infant mice during and Dixit, V.D. (2009). Obesity accelerates thymic aging. Blood 114,
chronic undernutrition. Science 351, 854–857. 3803–3812.

714 Cell Host & Microbe 23, June 13, 2018


Cell Host & Microbe

Review
Yatsunenko, T., Rey, F.E., Manary, M.J., Trehan, I., Dominguez-Bello, M.G., Personalized nutrition by prediction of glycemic responses. Cell 163,
Contreras, M., Magris, M., Hidalgo, G., Baldassano, R.N., Anokhin, A.P., 1079–1094.
et al. (2012). Human gut microbiome viewed across age and geography. Na-
ture 486, 222–227. Zhao, L., Zhang, F., Ding, X., Wu, G., Lam, Y.Y., Wang, X., Fu, H., Xue, X., Lu,
C., Ma, J., et al. (2018). Gut bacteria selectively promoted by dietary fibers alle-
Ze, X., Duncan, S.H., Louis, P., and Flint, H.J. (2012). Ruminococcus bromii is a viate type 2 diabetes. Science 359, 1151–1156.
keystone species for the degradation of resistant starch in the human colon.
ISME J. 6, 1535–1543. Zou, J., Chassaing, B., Singh, V., Pellizzon, M., Ricci, M., Fythe, M.D., Kumar,
M.V., and Gewirtz, A.T. (2018). Fiber-mediated nourishment of gut microbiota
Zeevi, D., Korem, T., Zmora, N., Israeli, D., Rothschild, D., Weinberger, A., protects against diet-induced obesity by restoring IL-22-mediated colonic
Ben-Yacov, O., Lador, D., Avnit-Sagi, T., Lotan-Pompan, M., et al. (2015). health. Cell Host Microbe 23, 41–53.e4.

Cell Host & Microbe 23, June 13, 2018 715

You might also like